Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Balkan J Med Genet ; 20(1): 81-86, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28924544

ABSTRACT

Mutations in the mitochondrial (mt) genome that result in mt dysfunction, have long been proposed to play important roles in the pathogenesis of hepatocellular carcinoma (HCC). Among these, the common mtDNA 4977 bp deletion is one of the most frequent mutations observed in various cancers. To understand the relationship between the mtDNA 4977 bp deletion and HCC, we performed mutational screening for the presence of this deletion in 105 HCC patients and 69 unrelated healthy subjects. After nested-polymerase chain reaction (nested-PCR) amplification, we found that there were 10 patients carrying the mtDNA 4977 bp deletion, and this deletion was absent in control subjects. Moreover, HCC patients carrying this deletion showed a marked increase in reactive oxygen species (ROS) level and mtDNA copy number when compared with the healthy controls. Taken together, our data indicated that the mtDNA 4977 bp deletion may play important role in the carcinogenesis of HCC, possibly via the alternation of mtDNA copy number and oxidative stress.

3.
Cancer Gene Ther ; 20(6): 342-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23661042

ABSTRACT

Genetically engineered tumor-selective vaccinia virus (VV) has been demonstrated to be a highly effective oncolytic agent, but immune clearance may limit its therapeutic potential. As previously demonstrated, immunosuppression can lead to significant enhancement of viral recovery and therapeutic effect, but the magnitude of complement-mediated viral inactivation has not been fully elucidated and warrants further investigation. Using fluorescent microscopy and quantitative plaque assays, we have determined complement's key role in viral clearance and its multi-faceted means to pathogen destruction. Complement can lead to direct viral destruction and inhibition of viral uptake into cells, even in the absence of anti-vaccinia antibodies. Our data demonstrate C5 to be integral to the clearance pathway, and its inhibition by Staphylococcal superantigen-like protein leads to a 90-fold and 150-fold enhancement of VV infectivity in both the presence and absence of anti-VV antibodies, respectively. This study suggests that complement inhibition may reduce vaccinia viral neutralization and may be critical to future in vivo work.


Subject(s)
Complement C5/antagonists & inhibitors , Immunosuppression Therapy , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Cell Line , Humans , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Vaccinia virus/immunology
4.
Cancer Gene Ther ; 19(3): 192-201, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22116377

ABSTRACT

Tumor necrosis factor superfamily members, including Fas ligand and TRAIL, have been studied extensively for cancer therapy, including as components of gene therapy. We examined the use of FasL expression to achieve tumor-selective replication of an oncolytic poxvirus (vFasL), and explored its biology and therapeutic efficacy for FasR- and FasR+ cancers. Infection of FasR+ normal and MC38 cancer cells by vFasL led to abortive viral replication owing to acute apoptosis and subsequently showed both reduced pathogenicity in non-tumor-bearing mice and reduced efficacy in FasR+ tumor-bearing mice. Infection of FasR- B16 cancer cells by vFasL led to efficient viral replication, followed by late induction of FasR and subsequent apoptosis. Treatment with vFasL as compared with its parental virus (vJS6) led to increased tumor regression and prolonged survival of mice with FasR- cancer (B16) but not with FasR+ cancer (MC38). The delayed induction of FasR by viral infection in FasR- cells provides for potential increased efficacy beyond the limit of the direct oncolytic effect. FasR induction provides one mechanism for tumor-selective replication of oncolytic poxviruses in FasR- cancers with enhanced safety. The overall result is both a safer and more effective oncolytic virus for FasR- cancer.


Subject(s)
Fas Ligand Protein/genetics , Oncolytic Virotherapy/methods , Poxviridae/physiology , fas Receptor/biosynthesis , Animals , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Breast Neoplasms/virology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Colorectal Neoplasms/virology , Fas Ligand Protein/biosynthesis , Fas Ligand Protein/metabolism , Female , Genetic Therapy , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Nude , Poxviridae/genetics , Virus Replication , fas Receptor/genetics
5.
Gene Ther ; 17(12): 1465-75, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20703311

ABSTRACT

Pre-existing antipoxvirus immunity in cancer patients presents a severe barrier to poxvirus-mediated oncolytic virotherapy. We have explored strategies of immunosuppression (IS) and/or immune evasion for efficient delivery of an oncolytic double-deleted vaccinia virus (vvDD) to tumors in the pre-immunized mice. Transient IS using immunosuppressive drugs, including tacrolimus, mycophenolate mofetil and methylprednisolone sodium succinate, have been used successfully in organ transplantation. This drug cocktail alone did not enhance viral recovery from subcutaneous tumor after systemic viral delivery. Using B-cell knockout mice, we confirmed that the neutralizing antibodies had a significant role in preventing poxvirus infection. Using a MC38 peritoneal carcinomatosis model, we found that the combination of IS and tumor cells as carriers led to the most effective viral delivery, viral replication and viral spread inside the tumor mass. We found that our immunosuppressive drug cocktail facilitated recruitment of tumor-associated macrophages and conversion into an immunosuppressive M2 phenotype (interleukin (IL)-10(hi)/IL-12(low)) in the tumor microenvironment. A combination of IS and carrier cells led to significantly prolonged survival in the tumor model. These results showed the feasibility of treating pre-vaccinated patients with peritoneal carcinomatosis using an oncolytic poxvirus and a combined immune intervention strategy.


Subject(s)
Immunosuppressive Agents/pharmacology , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Vaccinia virus/physiology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/immunology , Carcinoma/drug therapy , Cell Line, Tumor , Female , Haplorhini , HeLa Cells , Humans , Immunosuppressive Agents/analysis , Immunosuppressive Agents/therapeutic use , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Peritoneal Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
6.
Gene Ther ; 17(4): 550-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20182517

ABSTRACT

We have explored a unique combination therapy for metastatic colorectal cancer. This strategy combines a potent and new oncolytic poxvirus expressing a membrane-bound tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or TNFSF10) and oxaliplatin (Ox) chemotherapy. We hypothesized that TRAIL expression would increase the efficacy of the oncolytic poxvirus, and that the therapeutic efficacy would be further enhanced by combination with chemotherapy. The cytotoxicity to cancer cells by Ox, oncolytic vaccinia virus (VV) and trail gene-armed VV alone or in combination was tested in vitro. The trail gene armed oncolytic VV-expressed high levels of TRAIL in infected cancer cells and had greater potency as a cytotoxic agent compared with the parent VV. Ox alone exerted concentration-dependent cytotoxicity. In vitro, the combination of the two agents applied at suboptimal concentrations for individual therapy displayed synergy in inducing cancer cells into enhanced levels of apoptosis/necrosis. Western blot analyses were consistent with the notion that TRAIL induced cancer cell death mainly through apoptosis, whereas Ox and vJS6 induced cell death more through non-apoptotic death pathways. In two aggressive colorectal carcinomatosis models derived from human HCT116 and murine MC38 cells, the combination therapy displayed synergistic or additive antitumor activity and prolonged the survival of the tumor-bearing mice compared with either Ox chemotherapy or vvTRAIL-mediated oncolytic gene therapy alone. This combination strategy may provide a new avenue to treating peritoneal carcinomatosis and other types of metastases of colorectal cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Carcinoma/therapy , Colorectal Neoplasms/therapy , Genetic Therapy/methods , Organoplatinum Compounds/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Blotting, Western , Carcinoma/drug therapy , Carcinoma/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Flow Cytometry , Humans , Mice , Oxaliplatin , Poxviridae , Transfection
7.
Int J Immunogenet ; 37(1): 33-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19930406

ABSTRACT

There are reports of IL-1 complex gene polymorphisms in ankylosing spondylitis (AS; MIM 106300), but the results have been inconsistent among populations. Moreover, few studies examine the association between IL-1 complex gene polymorphisms and clinical symptoms of AS patients. We investigated polymorphisms of IL-1 complex with AS in the Chinese Han population in this study. Chinese Han AS patients and ethnically matched healthy controls were genotyped for five single nucleotide polymorphisms (IL1beta+3953, beta-511, F10.3, RN.4, RN.6/1) and the IL1RN.VNTR of IL-1 gene cluster. Allele, Genotype and haplotype frequencies were compared between cases and controls by SHEsis software. The frequency of allele C of the marker IL1F10.3 was significantly increased in AS patients versus controls [p = 0.001, odds ratio (OR) = 1.54, 95% confidence interval (CI) = 1.19-1.20; p = 0.002, respectively]. Strong linkage disequilibrium was identified between IL1B-511, IL1B+3953 and RN4 in both patients and healthy controls (D' > 0.95). Haplotypes of pairs of these markers (6) were also significantly associated with AS. The strongest associations observed was between allele combination B-511-T/B+3953-C/F10.3-C/RN4-T/RN2VNTR-1/RN6.1-C and AS (p = 3.32 x 10(-5), OR = 4.41, 95% CI=2.1-9.3). Clinical manifestation showed week association between RN2VNTR A2 allele and risk of peripheral arthritis (OR = 0.2, 95% CI = 0.07-0.91). The IL-1 gene cluster is associated with AS in Chinese population. This finding provides strong statistical support for the previously observed relationship and indicates possible association between clinical manifestation and genetic factor.


Subject(s)
Asian People/genetics , Ethnicity/genetics , Genetic Predisposition to Disease , Interleukin-1/genetics , Multigene Family/genetics , Spondylitis, Ankylosing/genetics , Adolescent , Adult , Case-Control Studies , Female , Gene Frequency/genetics , Haplotypes/genetics , Humans , Linkage Disequilibrium/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Young Adult
8.
Cancer Gene Ther ; 15(9): 616-24, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18535619

ABSTRACT

Vaccinia virus has recently been used as an expression vector for gene delivery and an oncolytic agent for cancer therapy. Although it has been established that interferon-induced double-stranded RNA (dsRNA)-activated protein kinase (PKR) and RNase L interfere with viral replication, little else is known about the other host factors that might affect viral replication and virus-mediated host cell killing. In this study, we evaluated the roles of c-Jun NH2-terminal kinase (JNK) in oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. We found that JNK knockout mouse embryonic fibroblasts (MEFs) were more susceptible to oncolytic vaccinia virus infection than wild-type MEFs. Moreover, viral replication and the production of infectious viral progeny were up to 100-fold greater in JNK-deficient MEFs than in wild-type MEFs. A similar result was observed for wild-type vaccinia virus. The increased killing of infected cells and the production of viral progeny was also observed in wild-type MEFs that had been treated with JNK inhibitors and in human colon cancer cells that had been transfected with dominant-negative JNK constructs. Moreover, testing on several human lung cancer cell lines and HeLa cells showed an inverse correlation between levels of JNK expression and susceptibility to oncolytic vaccinia virus. Our study also revealed that oncolytic virus infection-mediated PKR activation was blocked or diminished in JNK-deficient MEFs. The adenovirus-mediated ectopic expression of human PKR in JNK-deficient MEFs reduced vaccinia virus replication to the levels observed in wild-type MEFs, indicating that JNK is required for vaccinia virus to efficiently activate PKR. Our results demonstrated that the cellular status of JNK function can dramatically affect oncolytic vaccinia virus replication and vaccinia virus-mediated host cell killing. This finding may enable further improvements in oncolytic virotherapy using vaccinia virus.


Subject(s)
Mitogen-Activated Protein Kinase 9/genetics , Oncolytic Viruses/physiology , Vaccinia virus/physiology , Virus Replication , eIF-2 Kinase/metabolism , Animals , Cell Line, Tumor , Enzyme Activation/genetics , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 8/genetics , Oncolytic Viruses/genetics , Phosphorylation , RNA, Double-Stranded/metabolism , Vaccinia virus/genetics , eIF-2 Kinase/genetics
9.
Cancer Gene Ther ; 15(2): 115-25, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18084242

ABSTRACT

In this study, we assessed the ability of a highly tumor-selective oncolytic vaccinia virus armed with a yeast cytosine deaminase gene to infect and lyse human and murine ovarian tumors both in vitro and in vivo. The virus vvDD-CD could infect, replicate in and effectively lyse both human and mouse ovarian cancer cells in vitro. In two different treatment schedules involving either murine MOSEC or human A2780 ovarian carcinomatosis models, regional delivery of vvDD-CD selectively targeted tumor cells and ovarian tissue, effectively delaying the development of either tumor or ascites and leading to significant survival advantages. Oncolytic virotherapy using vvDD-CD in combination with the prodrug 5-fluorocytosine conferred an additional long-term survival advantage upon tumor-bearing immunocompetent mice. These findings demonstrate that a tumor-selective oncolytic vaccinia combined with gene-directed enzyme prodrug therapy is a highly effective strategy for treating advanced ovarian cancers in both syngeneic mouse and human xenograft models. Given the biological safety, tumor selectivity and oncolytic potency of this armed oncolytic virus, this dual therapy merits further investigation as a promising new treatment for metastatic ovarian cancer.


Subject(s)
Carcinoma/therapy , Cytosine Deaminase/genetics , Oncolytic Virotherapy , Ovarian Neoplasms/therapy , Saccharomyces cerevisiae/genetics , Vaccinia virus/genetics , Virus Replication , Animals , Antimetabolites/administration & dosage , Antimetabolites/therapeutic use , Carcinoma/drug therapy , Cell Line, Tumor , Combined Modality Therapy , Cytosine Deaminase/administration & dosage , Cytosine Deaminase/therapeutic use , Female , Flucytosine/administration & dosage , Flucytosine/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Ovarian Neoplasms/drug therapy , Saccharomyces cerevisiae/enzymology , Vaccinia virus/physiology , Virus Replication/genetics
10.
Gene Ther ; 14(8): 638-47, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17268533

ABSTRACT

To enhance further the safety and efficacy of oncolytic vaccinia virus, we have developed a new virus with targeted deletions of three viral genes encoding thymidine kinase and antiapoptotic/host range proteins SPI-1 and SPI-2 (vSPT). Infection of human and murine tumor cell lines yielded nearly equivalent or a log lower virus recovery in comparison to parental viruses. Viral infection activated multiple caspases in cancer cells but not in normal cells, suggesting infected cells may die via different pathways. In tumor-bearing mice, vSPT recovery from MC38 tumor was slightly reduced in comparison to two parental viruses. However, no virus was recovered from the brains and livers of mice injected with vSPT in contrast to control viruses. vSPT demonstrated significantly lower pathogenicity in nude mice. Systemic delivery of vSPT showed significant tumor inhibition in subcutaneous MC38 tumor, human ovarian A2780 and murine ovarian MOSEC carcinomatosis models; however, the tumor inhibition by vSPT was reduced compared with parental viruses. These results demonstrated that although deletion of these three viral genes further enhanced tumor selectivity, it also weakened the oncolytic potency. This study illustrates the complexity of creating a tumor-selective oncolytic virus by deleting multiple viral genes involved in multiple cellular pathways.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Vaccinia virus/genetics , Animals , Cell Line, Tumor , Female , Gene Deletion , Gene Targeting/methods , Genetic Engineering , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Models, Animal , Neoplasm Transplantation , Protein Kinases/genetics , Recombinant Fusion Proteins/genetics , Safety , Sp2 Transcription Factor/genetics , Thymidine Kinase/genetics , Transplantation, Heterologous , Virus Replication
11.
Gene Ther ; 9(3): 168-75, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11859419

ABSTRACT

Transcriptional targeting of gene expression has been plagued by the weakness of tissue-specific promoters. Thus, to increase promoter strength while maintaining tissue specificity, we constructed a recombinant adenovirus containing a binary promoter system with a tumor-specific promoter (CEA; carcinoembryonic antigen) driving a transcription transactivator, which then activates a minimal promoter to express a suicide gene (HSV-tk; herpes simplex virus thymidine kinase). This ADV/binary-tk induced equal or greater cell killing in a CEA-specific manner in vitro compared with the CEA-independent killing of a vector with a constitutive viral promoter driving HSV-tk (ADV/RSV-tk). To monitor adenovirus-mediated HSV-tk gene expression in vivo, we employed noninvasive nuclear imaging using a radioiodinated nucleoside analog ([((1)31)I]-FIAU) serving as a substrate for HSV-tk. [((1)31)I]-FIAU-derived radioactivity accumulated after intratumoral injection of ADV/binary-tk only in the area of CEA-positive tumors with significantly less spread to the adjacent liver tissue than after administration of the universally expressed ADV/RSV-tk. Both viruses exhibited similar antitumor efficacy upon injection of liver metastases. Importantly, in vivo dose escalation studies demonstrated significantly reduced toxicity after intravenous administration of ADV/binary-tk versus ADV/RSV-tk. In summary, the increased therapeutic index of this novel, amplified CEA-driven suicide gene therapy vector is a proof of principle for the powerful enhancement of a weak tissue-specific promoter for effective tumor restricted gene expression.


Subject(s)
Breast Neoplasms/therapy , Carcinoembryonic Antigen/genetics , Gene Targeting/methods , Genetic Therapy/methods , Transcription, Genetic , Adenoviridae/genetics , Animals , Gene Expression , Genetic Vectors/administration & dosage , HeLa Cells , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Injections, Intralesional , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , Retroviruses, Simian/enzymology , Simplexvirus/enzymology , Thymidine Kinase/genetics , Tumor Cells, Cultured
12.
J Immunother ; 24(2): 151-61, 2001.
Article in English | MEDLINE | ID: mdl-11265773

ABSTRACT

Global alterations in chromatin structure profoundly influence gene expression in thoracic neoplasms, silencing tumor suppressors while facilitating the expression of various cancer testis antigens such as NY-ESO-1. Although recent studies have shown that histone deacetylase inhibitors can potentiate tumor suppressor gene induction mediated by demethylating agents in cancer cells, the ability of these agents to augment cancer testis antigen expression have not been fully defined. The authors designed the current study to determine whether the histone deacetylase inhibitor, depsipeptide FR901228 (DP), could enhance NY-ESO-1 induction mediated by the DNA demethylating agent 5-Aza-2'-deoxycytidine (DAC) in cell lines established primarily from thoracic cancers. Quantitative reverse-transcriptase polymerase chain reaction analysis revealed that, under exposure conditions potentially achievable in clinical settings, DAC dramatically induced NY-ESO-1 expression in cultured cancer lines. DP alone mediated negligible target gene induction but significantly augmented DAC-mediated induction of NY-ESO-1. After DAC or sequential DAC-DP treatment, HLA-A*0201 cancer cells were recognized by an HLA-A*0201 CTL specific for NY-ESO-1. Although sequential DAC/DP exposure did not uniformly enhance immune recognition of target cells compared with DAC alone, this treatment mediated profound induction of apoptosis in cancer cells but not normal human bronchial epithelia. The apoptotic effects of DAC, DP, or sequential DAC-DP did not correlate in an obvious manner with histology, or the magnitude of NY-ESO-1 induction in cancer cells. Although the mechanisms have not been fully defined, sequential DAC-DP treatment may be a novel strategy to augment antitumor immunity in cancer patients.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Apoptosis/drug effects , Depsipeptides , Membrane Proteins , Neoplasms/pathology , Peptides, Cyclic , Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antigens, Neoplasm/immunology , Blotting, Western , Breast Neoplasms , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Esophageal Neoplasms , Flow Cytometry , Humans , Lung Neoplasms , Melanoma , Mesothelioma , Neoplasms/drug therapy , Neoplasms/immunology , Pleural Neoplasms , Proteins/analysis , Tumor Cells, Cultured
13.
Mol Ther ; 3(3): 278-83, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11273768

ABSTRACT

Though extensively studied, the use of tissue- or cell-type-specific promoters to target transgene expression is hampered by their weak activity. We hypothesized that this problem could be addressed by using a GAL4 gene regulatory system, wherein a weak, tissue-specific promoter would drive expression of the GAL4/VP16 fusion protein (GV16), which in turn would transactivate a minimal synthetic promoter, GAL4/TATA (GT), upstream of a transgene. To test this hypothesis, we constructed adenoviral vectors expressing a lacZ or GV16 gene driven by a carcinoembryonic antigen (CEA) promoter (Ad/CEA-LacZ or Ad/CEA-GV16) and evaluated levels of transgene expression they produced in cultured cells and in subcutaneous tumors after intratumoral administration. In CEA-positive cells, treatment with Ad/CEA-GV16 + Ad/GT-LacZ versus Ad/CEA-LacZ increased transgene expression 20- to 100-fold. In CEA-negative cells, treatment with Ad/CEA-GV16 + Ad/GT-LacZ increased transgene expression to a much lower degree (6- to 8-fold). In addition, analysis of Bax gene-mediated cell death revealed that this system can be used to avoid Bax's toxic effects on CEA-negative cells without compromising its ability to kill CEA-positive cells in vitro and in vivo. Thus, the combination of a tissue-specific promoter with the GAL4 gene regulatory system could be useful for targeting transgene expression.


Subject(s)
Adenoviridae/genetics , Carcinoembryonic Antigen/genetics , Fungal Proteins/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2 , Saccharomyces cerevisiae Proteins , Transcription Factors/genetics , Transgenes , Animals , Cell Line , DNA-Binding Proteins , Fungal Proteins/pharmacology , Genes, Regulator , Genetic Therapy , HeLa Cells , Herpes Simplex Virus Protein Vmw65/genetics , Humans , Mice , Mice, Nude , Neoplasms/genetics , Proto-Oncogene Proteins , Recombinant Fusion Proteins/genetics , TATA Box , Transcription Factors/pharmacology , bcl-2-Associated X Protein
14.
Ann Thorac Surg ; 71(1): 295-301; discussion 301-2, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11216765

ABSTRACT

BACKGROUND: Although MAGE-3 has been detected in approximately 40% of lung and esophageal cancers, expression of this cancer testis antigen appears to be below the threshold for immune recognition in patients with these malignancies. The aim of this study was to determine if the demethylating agent, 5-Aza-2'-deoxycytidine (DAC) and if the histone deacetylase inhibitor Depsipeptide FR901228 (DP) could enhance MAGE-3 expression in lung and esophageal cancer cells. METHODS: Eleven lung and esophageal cancer lines and cultured normal human bronchial epithelial (NHBE) cells were exposed to normal media (NM), DAC, DP, or combination DAC/DP at varying concentrations and exposure durations. MAGE-3 expression was evaluated by quantitative RT-PCR (TaqMan) and immunohistochemistry techniques. Trypan blue exclusion techniques were used to examine the proliferation of cancer cells after drug exposure. RESULTS: Relative to untreated controls, MAGE-3 expression was enhanced 32-fold (range 3.9 to 110) by DAC alone (0.1 micromol/L x 72 h), 2.1-fold (0.4 to 4.2) by DP alone (25 ng/mL x 6h), and 57-fold (4.6 to 209) by sequential DAC/DP exposure. Increased MAGE-3 mRNA copy numbers coincided with enhanced protein levels in these cells. MAGE-3 expression persisted after drug exposure. Flow cytometry confirmed the presence of functional HLA class I expression in these cells. Sequential DAC/DP treatment mediated pronounced growth inhibition in cancer cells but not NHBE. CONCLUSIONS: Sequential DAC/DP treatment may be a novel strategy to simultaneously augment MAGE-3 expression and induce growth arrest in thoracic malignancies.


Subject(s)
Antigens, Neoplasm/metabolism , Azacitidine/analogs & derivatives , Depsipeptides , Esophageal Neoplasms/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Peptides, Cyclic , Adenocarcinoma/metabolism , Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Azacitidine/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , Immunohistochemistry , Melanoma/metabolism , Neoplasm Proteins/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects
15.
Nat Med ; 6(7): 826-31, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888935

ABSTRACT

Medulloblastoma is the most malignant pediatric brain tumor. It is believed to originate from the undifferentiated external granule layer cells in the cerebellum, but the mechanism of tumorigenesis remains unknown. Here we studied three types of human medulloblastoma cells that express markers corresponding to different levels of neuronal differentiation. They expressed the neuronal repressor element 1 (RE1) silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF; refs. 7-10) at very high levels compared with either neuronal progenitor NTera2 (NT2) cells or fully differentiated human neuron teratocarcinoma (hNT cells). To counter the effect of REST/NRSF, we used a recombinant transcription factor, REST-VP16, constructed by replacing repressor domains of REST/NRSF with the activation domain of viral protein (VP16). Transient expression of REST-VP16 in medulloblastoma cells was able to compete with the endogenous REST/NRSF for DNA binding and stimulate neuronal promoters. High-efficiency expression of REST-VP16 mediated by adenovirus vectors (Ad.REST-VP16) in medulloblastoma cells was able to counter REST/NRSF-mediated repression of neuronal promoters, stimulate expression of endogenous neuronal genes and trigger apoptosis through the activation of caspase cascades. Furthermore, intratumoral injection of Ad.REST-VP16 in established medulloblastoma tumors in nude mice inhibited their growth. Therefore, REST/NRSF may serve as a new target for therapeutic interventions for medulloblastoma through agents such as REST-VP16.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Neurons/cytology , Repressor Proteins/metabolism , Transcription Factors , Adenoviridae/genetics , Animals , Apoptosis , Cell Differentiation , Gene Expression Regulation, Neoplastic , Herpes Simplex Virus Protein Vmw65/genetics , Herpes Simplex Virus Protein Vmw65/metabolism , Humans , Mice , Mice, Nude , Neoplasms, Experimental/therapy , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Repressor Proteins/genetics , Tumor Cells, Cultured
16.
Se Pu ; 18(1): 1-4, 2000 Jan.
Article in Chinese | MEDLINE | ID: mdl-12541442

ABSTRACT

This paper used artificial neural network (ANN) modeling method to study the relationship between the column efficiency and the operating conditions. This method solved the problem that it is not easy to establish a quantitative model between the column efficiency and its main effecting factors using those traditional modeling methods, as the relationship is usually quite complex and non-linear in fact. The varied-pace BP (back-propagation) learning algorithm was adopted, and a three-layer weight-connected ANN model for a typical dual column system was established. The effective plate number representing the column efficiency acted as the output vectors, while the temperature of the pre-column, the temperature of the main column, the pressure difference between the columns and the vent rate acted as the input vectors. Then the model acquired was used to predict column efficiency (characterized by "effective plate number") under different operating conditions. The results showed that the model predicting value was in consistent with the value found. This work proved that ANN modeling method was suitable for the study on the relationship between the column efficiency of two-dimensional column chromatography system and the operating conditions.


Subject(s)
Chromatography/methods , Neural Networks, Computer , Algorithms , Forecasting
17.
Cancer Res ; 59(20): 5186-93, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10537296

ABSTRACT

The feasibility of noninvasive imaging of adenoviral-mediated herpes virus type one thymidine kinase (HSV1-tk) gene transfer and expression was assessed in a well-studied animal model of metastatic colon carcinoma of the liver. Tumors were produced in syngeneic BALB/c mice by intrahepatic injection of colon carcinoma cells (MCA-26). Seven days later, three different doses (3 x 10(8), 1 x 10(8), and 3 x 10(7) plaque-forming units (pfu) of the recombinant adenoviral vector ADV. Rous sarcoma virus (RSV)-tk bearing the HSV1-tk gene were administered by intratumoral injection in separate groups of mice. Two control groups of tumor-bearing mice received intratumoral injections of the control adenoviral vector dl-312 or buffer alone, respectively. T2-weighted magnetic resonance (MR) images of mice were obtained before administering the virus and provided an anatomical reference of hepatic tumor localization. Eighteen h after the virus injection, one group of animals was given i.v. injections of 300 microCi of no-carrier-added 5-[131I]-2'-fluoro-1-beta-D-arabinofuranosyluracil (FIAU) and imaged 24 h later with a gamma camera. In some animals, the tumors were sampled and processed for histology and quantitative autoradiography (QAR). The gamma camera images demonstrated highly specific localization of [131I]FIAU-derived radioactivity to the area of ADV.RSV-tk-injected tumors in the liver, which was confirmed by coregistering the gamma camera and T2-weighted MR images. There was no accumulation of [131I]FIAU-derived radioactivity in tumors that were injected with the control vector or injection solution alone. A more precise distribution of radioactivity in the area of transfected tumor was obtained by histological and QAR comparisons. A heterogeneous pattern of radioactivity distribution in transfected tumors was observed. A punctate pattern of radioactivity distribution was observed in peritumoral liver tissue in animals given injections of 3 x 10(8) and 1 x 10(8) pfu of ADV.RSV-tk but not in animals given injections of 3 x 10(7) pfu nor in control animals. A QAR-microscopic comparison showed that the punctate areas of radioactivity colocalized with cholangial ducts. The level of [131I]FIAU-derived radioactivity accumulation (HSV1-tk expression) in the transfected tumors was viral dose-dependent. The viral dose-dependency of radioactivity accumulation was more pronounced in peritumoral liver, which was confirmed by reverse transcription-PCR analysis. A separate group of tumor-bearing animals received different doses of ADV.RSV-tk vector followed by treatment with ganciclovir (GCV), 10 mg/kg i.p. b.i.d. for 6 days. The ADV.RSV-tk transfected tumors significantly regressed with GCV treatment; the control tumors continued to grow. During the GCV treatment, the levels of liver transaminases (ALT and AST) were significantly increased in animals that received injections of 3 x 10(8) and 1 x 10(8) pfu of ADV.RSV-tk but not in animals that received injections of 3 x 10(7) pfu and in control animals. The observed liver toxicity confirms the results of gamma camera and QAR imaging, which demonstrated an unwanted spread of ADV.RSV-tk vector and HSV1-tk expression in peritumoral and remote liver tissue at higher doses. These and our previous results indicate that noninvasive imaging of adenoviral-mediated HSV1-tk gene expression is feasible for monitoring cancer gene therapy in patients.


Subject(s)
Adenoviridae/genetics , Colonic Neoplasms/therapy , Gene Transfer Techniques , Genetic Therapy , Simplexvirus/enzymology , Thymidine Kinase/genetics , Animals , Arabinofuranosyluracil/analogs & derivatives , Autoradiography , Ganciclovir/therapeutic use , Gene Expression , Iodine Radioisotopes , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
18.
Cancer Res ; 59(24): 6068-73, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10626792

ABSTRACT

Although SV40 oncoproteins have been detected in malignant pleural mesotheliomas (MPMs), their role in the pathogenesis and clinical behavior of these neoplasms remains controversial. In the present study, we sought to define the relevance of SV40 T/t antigen expression in established human mesothelioma cell lines deficient for p16INK4a as well as ARF expression. SV40 early region sequences were readily detected in genomic DNA isolated from pleural mesothelioma lines; however, levels of SV40 T/t antigen expression were highly variable in these cells. An adenoviral vector expressing an antisense transcript to SV40 early region inhibited T antigen expression and mediated significant growth inhibition and apoptosis in T-antigen-positive mesothelioma cells and SV40-transformed COS-7 cells. Abrogation of T/t antigen expression coincided with enhanced p21/WAF-1 expression, suggesting that restoration of p53-mediated pathways may have contributed to the growth inhibition and apoptosis induced by the antisense construct. These effects were not observed after similar treatment of mesothelioma or lung cancer cells containing no SV40 DNA sequences. Collectively, these data suggest that SV40 oncoproteins contribute to the malignant phenotype of pleural mesotheliomas and indicate that interventions designed to abrogate their expression may be efficacious in the treatment of individuals with these neoplasms.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Apoptosis , Mesothelioma/drug therapy , Oligonucleotides, Antisense/therapeutic use , Pleural Neoplasms/drug therapy , Simian virus 40/genetics , Adenoviridae/genetics , Cell Division/drug effects , Gene Transfer Techniques , Genes, Immediate-Early , Genes, Viral , Genetic Vectors/genetics , Humans , Mesothelioma/metabolism , Oncogene Proteins/physiology , Phenotype , Pleural Neoplasms/metabolism , Tumor Cells, Cultured
19.
Gene Ther ; 3(9): 802-10, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8875229

ABSTRACT

Transgene expression in studies of both gene function and gene therapy may be assisted considerably through the use of transcriptional regulatory elements which permit high-level, and/or tissue-specific gene expression. We have therefore evaluated the transcriptional activities of a series of viral and cellular enhancer/promoter elements, both in vitro and in vivo. The five enhancer/promoter elements showing either high-level or hepatocyte-specific expression following transient transfection into hepatoma cells were incorporated into recombinant adenoviruses expressing human alpha 1-antitrypsin (hAAT) for in vivo studies in the liver of immunodeficient and immunocompetent mice. The human elongation factor 1 alpha gene promoter produced 2 mg/ml serum level of hAAT, which is physiologic in humans and will be therapeutic for patients with AAT deficiency. This and all other enhancer/promoters except that of the CMV-IE gene yielded persistent hAAT expression in SCID mice. These findings demonstrate that adenovirus vectors provide an effective system for studies designed to evaluate enhancer/promoter activities in vivo. Several of the enhancer/promoters examined in this study will have significant utility in adenovirus-mediated gene therapy for alpha 1-antitrypsin deficiency and other genetic disorders.


Subject(s)
Adenoviruses, Human/genetics , Gene Expression , Gene Transfer Techniques , Liver/metabolism , Promoter Regions, Genetic/genetics , Animals , Carcinoma, Hepatocellular , DNA, Viral/analysis , Enhancer Elements, Genetic/genetics , Female , Genetic Vectors/genetics , Humans , Liver Neoplasms , Mice , Mice, Inbred C57BL , Mice, SCID , Organ Specificity , Peptide Elongation Factor 1 , Peptide Elongation Factors/genetics , RNA, Messenger/analysis , Tumor Cells, Cultured , alpha 1-Antitrypsin/analysis , alpha 1-Antitrypsin/genetics
20.
Zhonghua Nei Ke Za Zhi ; 32(4): 243-5, 1993 Mar.
Article in Chinese | MEDLINE | ID: mdl-8156850

ABSTRACT

We studied the change of the binding capacity of the sex hormone binding globulin (SHBG-BC) and correlated it with the serum level of thyroid hormone and sex hormone in 17 patients (10 males and 7 female) with hyperthyroidism before and during the 16 weeks of antithyroid treatment. The serum TT4, TT3, FT4I level and SHBG-BC were significantly elevated before treatment compared to normal adults. After the treatment with antithyroid drugs, serum SHBG-BC decreased significantly at 2nd weeks in female (from 312.9 +/- 39.6 to 205 +/- 18.6 nmol/L, P < 0.05) and at 8th weeks in male (from 155.7 +/- 18.6 to 109.7 +/- 7.9 nmol/L, P < 0.05). It continued to decrease to normal range (78.6 +/- 7.3 vs 65.0 +/- 24.1 nmol/L, P > 0.05) at 8th weeks in female, but was still higher than normal range (107.4 +/- 7.2 vs 41.5 +/- 10.2 nmol/L, P < 0.001) even at 16th weeks in male. The change of SHBG-BC was significantly positively correlated to the serum concentration of TT4, TT3 and FT4I(P < 0.001). In male patients the serum testosterone (T) level decreased from a high level of 41.9 +/- 6.2 nmol/L before treatment to 25.4 +/- 3.4 nmol/L at 8th weeks (P < 0.05) and to 19.8 +/- 2.8 nmol/L at 16 weeks which was in normal range. The decrease of serum T level was also positively correlated to the changes of SHBG-BC (P < 0.0001). The serum estradiol (E2) level of female patients was in the upper normal range before the antithyroid treatment.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Hyperthyroidism/drug therapy , Hyperthyroidism/metabolism , Methimazole/therapeutic use , Sex Hormone-Binding Globulin/metabolism , Adult , Estradiol/blood , Female , Humans , Male , Prospective Studies , Testosterone/blood , Thyroxine/blood , Triiodothyronine/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...